Download Allogeneic haematopoietic stem cell transplantation

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts
no text concepts found
Transcript
Bone Marrow Transplantation (2006) 38, 1–4
& 2006 Nature Publishing Group All rights reserved 0268-3369/06 $30.00
www.nature.com/bmt
EDITORIAL
Allogeneic haematopoietic stem cell transplantation for severe autoimmune
diseases: great expectations but controversial evidence
Bone Marrow Transplantation (2006) 38, 1–4.
doi:10.1038/sj.bmt.1705394
Autoimmune diseases (ADs) have been defined as a
‘fascinating but still poorly understood group of diseases’,
which pose ‘some of the most baffling scientific questions
and daunting clinical challenges in internal medicine’.1 This
statement still holds true, and it is generally recognized that
ADs result from three interacting components: genetic,
environmental and regulatory.2,3 In spite of this complexity
and of considerable overlap, there is a tendency to
distinguish ADs with an impaired immune system, also
known as primary ADs, from those that are predominantly
antigen driven.4 There is no better demonstration of the
latter than the clinical studies that have shown the
disappearance of autoantibodies (antithyroid, anti-tissue
transglutaminase) following removal of the antigens (by
thyroidectomy or by gluten-free diet, respectively).5,6 The
proposal that transplantation with genetically engineered
stem cells with the culprit antigen expressed by donorantigen-presenting cells in the thymus might result in
immunological tolerance7 is in accordance with this current
line of thought, which includes the Matzinger ‘danger’
model of autoimmunity.8,9 A different outlook is apparent
for the primary ADs, in which profound abnormalities of
the patients’ immune system are in the foreground.
Systemic lupus erythematosus (SLE) is the most prominent
example of this type of pathogenetic mechanism, where the
intrinsic tendency of B cells to respond excessively to
immune stimulation is thought to be an essential feature of
the disease,10 and causes a veritable ‘autoantibody explosion’.11
Following pioneering experimental studies,12,13 autologous haematopoietic stem cell transplantation (autoSCT)
has now become an accepted therapeutic procedure for
severe ADs, despite the paradox of trying to restore
tolerance by transplanting the patients’ own stem cells.
Extensive reviews have been published, some very recently.14–17 Even if there is still some uncertainty as to
whether genuine tolerance may be really achieved,18,19
disease progression has been clearly shown to be delayed,
especially following high-dose conditioning, albeit with the
drawback of some transplant-related mortality (TRM).20
In order to reduce TRM to a minimum, more specifically
lymphoablative conditioning regimens have been proposed.21 However, in spite of its accepted therapeutic
significance, of the long-term remissions that can be
sometimes achieved22,23 and of the capacity to respond to
treatment again in previously refractory patients, no
authentic cure can be expected.24–26 This is well exemplified
by two patients with coexisting AD and malignant disease
in whom autoSCT was followed by cure of the malignancy
but not of the AD.27,28
The suggestion that allogeneic SCT (alloSCT) might
have a more far-reaching effect was based originally on the
favourable results in patients with AD coexisting with other
diseases.29 A position paper was published recently that
stated that alloSCT was expected to be highly effective for
inducing sustained remissions or ‘cure’ of autoimmune
diseases.30 A series of mechanisms were considered,
including immunomodulation, tolerization by regulatory
T cells and, most importantly, immune-mediated destruction of autoreactive cells.31 By analogy with the multiple
clinical diversifications of immune cellular immunotherapy,32 this last effect was defined as graft-versus-autoimmunity (GVA).33,34 As with the more common and better
known graft-versus-leukaemia effect, also GVA was found
to be more robust when associated with graft-versus-host
disease.35,36
On the other hand, it has been claimed that mixed
chimaerism might be sufficient to keep under control the
autoreactive mechanisms. This is certainly true in experimental autoimmunity, where it has been investigated and
confirmed in numerous laboratories.37–39 However, the
situation is far from being so straightforward in clinical
medicine. Here, along with cases in which post-alloSCT
mixed chimaerism proved effective in controlling the
AD,40,41 there are others in which it was accompanied by
relapse of AD.42,43 The concept that complete remission of
AD depends upon full donor chimaerism has been
validated by the favourable effects of donor lymphocyte
transfusions (DLI) post transplant, designed to obtain full
donor chimaerism.44–46 Unfortunately, there are also cases
in which the AD relapsed in spite of full donor chimaerism.
The first is the now famous patient with rheumatoid
arthritis (RA) in Toronto, who underwent alloSCT from
her HLA-identical brother because of gold-induced aplasia,
and then relapsed with RA 2 years later despite continuing
full donor chimaerism.47 Chimaerism was assessed by
cytogenetics and by tandem repeat-based DNA typing. In
the other case of two concomitant diseases, a patient with
seropositive RA received a nonmyeloablative allogeneic
peripheral blood SCT because of multiple myeloma,
achieved complete remission of both diseases with disappearance of paraprotein and rheumatoid factor (RF), but
relapsed with RA 10 months after transplantation.48 It may
be noted that the HLA-identical brother donor had weak
RF positivity. In addition, peripheral blood transplants are
devoid of mesenchymal cells, which may have a beneficial
effect on AD.49 Perhaps the most informative case of AD
Editorial
2
relapsing in spite of full donor chimaerism has been
recently observed in Genoa. This was a patient with
refractory Evans syndrome who had been transplanted
with bone marrow from his HLA-identical sister in 2000,
and had received five DLIs in order to achieve full donor
chimaerism and complete clinical and haematologic remission.50 However, he had a catastrophic relapse 5 years later.
Full donor chimaerism persisted, and the supernatants of
ex vivo-cultured and expanded B lymphocytes contained
immunoglobulin (Ig)G and IgM that did not react with
the panel of erythrocytes against which the antibodies
eluted from the patient’s Coombs-positive red cells51 were
directed.
The immunologic interpretation of these almost paradoxical relapses in patients with full post transplant donor
chimaerism is still obscure. T cells appear to be involved in
the RA relapses, and it has been hypothesized that residual
recipient cells could sensitize HLA-identical donor T cells
to perpetuate the immunologic imbalance underlying RA.48
However, in a typical B-mediated disease such as Evans
syndrome, a more appropriate interpretation could originate from the notion of long-lived recipient plasma cells,
the contribution of which to long-standing, refractory
humoral autoimmunity has been shown recently.52,53 Shortlived plasma blasts have been shown to become long-lived
plasma cells, occupying postulated survival niches. In NZB/
W mice, a typical model for SLE, long-lived autoreactive
plasma cells have been demonstrated in the bone marrow
and in the spleen.54 However, it is a classical notion that
they can be eradicated following alloSCT, a finding that
was pivotal for the development of stem cell therapy for
autoimmunity in humans.55 An array of cytokines are
necessary for plasma cell survival, such as the well-known
interleukin-6 and also BAFF and APRIL,56 but the most
relevant for plasma cell maintenance has been shown to be
Blimp-1.57 Allotransplantation in lupus mice has been
shown to overcome the disease,12–14 but, in the light of these
observations, one could suspect that a greater degree of
refractoriness may be found in human patients with
systemic autoimmune diseases than suspected previously.
Single case reports, no matter how carefully studied,
cannot provide the final answer but still they should not be
ignored. If it were confirmed that minimal residual
autoimmune disease is capable of surviving the GVA
effects of alloSCT, then the possible advantage of alloSCT
over the safer, although perhaps immunologically less
appealing, autoSCT procedure would inevitably be reduced.
AM Marmont
Division of Haematology and Stem Cell Transplantation
Centre, S Martino’s Hospital, Genoa, Italy
E-mail: [email protected]
References
1 Davidson A, Diamond B. Autoimmune diseases. N Engl J Med
2001; 345: 340–350.
2 Ermann J, Fathman CG. Autoimmune diseases: genes, bugs
and failed regulation. Nat Immunol 2002; 2: 759–766.
Bone Marrow Transplantation
3 Fathman CG, Soares L, Chan SM, Utz PJ. An array of
possibilities for the study of autoimmunity. Nature 2005; 435:
605–611.
4 Amital H, Shoenfeld Y. Autoimmunity and autoimmune
diseases such as systemic lupus erythematosus. In: Lahita
RG (ed). Systemic Lupus Erythematosus, 4th edn. Elsevier:
Amsterdam-Tokyo, 2004, pp 3–27.
5 Chiovato L, Latrofa F, Braverman LE, Pacioni F, Capezzone
M, Masserini L et al. Disappearance of humoral thyroid
autoimmunity after complete removal of thyroid antigens. Ann
Intern Med 2003; 139: 346–351.
6 Ventura A, Not T, Tommasini A, Marzari R, Sblattero D.
Autoantibodies as predictors of disease. Lancet 2004; 364:
1403.
7 Alderuccio F, Murphy K, Toh B-H. Stem cells engineered to
express self-antigen to treat autoimmunity. Trends Immunol
2003; 24: 176–180.
8 Matzinger P. Tolerance, danger and the extended family. Annu
Rev Immunol 1994; 12: 991–1045.
9 Reines BP. Bystanders or bad seeds? Many autoimmune-target
cells may be transforming in cancer and signaling ‘danger’ to
the immune system. Autoimmunity 2001; 33: 121–134.
10 Lipsky PE. Systemic lupus erythematosus: an autoimmune
disease of B cell hyperreactivity. Nat Immunol 2002; 2:
764–766.
11 Sherer Y, Gorstein A, Fritzler M, Shoenfeld Y. Autoantibody
explosion in systemic lupus erythematosus: more than 100
different antibodies found in SLE patients. Semin Arthritis
Rheum 2004; 34: 501–537.
12 Ikehara S. Treatment of autoimmune disease by hematopoietic
stem cell transplantation. Exp Hematol 2001; 29: 661–669.
13 van Bekkum DW. Preclinical experiments. Best Pract Res Clin
Haematol 2004; 17: 201–222.
14 Burt RK, Marmont AM. Stem Cell Therapy for Autoimmune
Disease. Landes Bioscience: Georgetown, TX, USA, 2004.
15 Burt RK, Slavin S, Burns W, Marmont AM. Induction of
tolerance in autoimmune diseases by hematopoietic stem cell
transplantation. Getting closer to a cure? Blood 2002; 99:
768–784.
16 Hough RE, Snowden JA, Wulffraat NM. Haematopoietic
stem cell transplantation in autoimmune diseases: a European
perspective. Br J Haematol 2005; 128: 432–459.
17 Tyndall A, Saccardi R. Haematopoietic stem cell transplantation in the treatment of severe autoimmune disease: results
from phase I/II studies, prospective randomized trials and
future directions. Clin Exp Immunol 2005; 141: 1–9.
18 Sun W, Popat U, Hutton G, Ying CQ, Zhang J, Krance R
et al. Characteristics of T-cell receptor repertoire and myelinreactive T cells reconstituted from autologous haematopoietic
stem-cell grafts in multiple sclerosis. Brain 2004; 127:
996–1008.
19 Muraro P, Douek DK, Packer A, Chung K, Guenaga FJ,
Cassini-Ingoni R et al. Thymic output generates a new and
diverse TCR repertoire after autologous stem cell transplantation in multiple sclerosis patients. J Exp Med 2005; 201:
805–816.
20 Gratwohl A, Passweg J, Bocelli-Tyndall C, Fassas A, van Laar
JM, Farge D et al. Autologous hematopoietic stem cell
transplantation for autoimmune diseases. Bone Marrow
Transplant 2005; 35: 869–879.
21 Burt RK, Oyama Y, Marmont AM, Slavin S, Arnold R, Hiepe
F et al. Randomized controlled trials of autologous stem cell
transplantation for autoimmune diseases: the evolution from
myeloablative to lymphoablative regimens. Arthritis Rheum
2006 (in press).
22 Burt RK, Traynor A, Statkute L, Barr WG, Rosa R,
Schroeder J et al. Nonmyeloablative hematopoietic stem cell
Editorial
3
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
transplantation for systemic lupus erythematosus. JAMA
2006; 295: 527–535.
Marmont AM, Gualandi F, van Lint MT, Guastoni C,
Bacigalupo A. Long-term complete remission of severe
nephrotic syndrome secondary to diffuse global (IV-6) lupus
nephritis following autologous, haematopoietic peripheral
stem (CD34+) transplantation. Lupus 2006; 15: 44–46.
Brenner MK. Haematopoietic stem cell transplantation for
autoimmune disease: limits and future potential. Best Pract
Res Clin Haematol 2004; 17: 359–374.
Petri M, Brodsky R. High-dose cyclophosphamide and stem
cell transplantation for refractory systemic lupus erythematosus. JAMA 2006; 295: 559–560.
Euler HH, Marmont AM, Bacigalupo A, Fastenrath S, Dreger
P, Hoffknecht M et al. Early recurrence or persistence of
autoimmune diseases after unmanipulated autologous stem
cell transplantation. Blood 1996; 88: 3621–3625.
Rosler W, Wanger B, Repp R, Kalden JR, Gramatzki M.
Autologous PBSCT in a patient with lymphoma and Sjogren
syndrome. Complete remission of lymphoma without control
of the autoimmune disease. Bone Marrow Transplant 1998; 22:
211–213.
Ferraccioli G, Damato R, De Vita S, Fanin R, Damiani D,
Baccarani M. Hematopoietic stem cell transplantation (HSCT)
in a patient with Sjogren’s syndrome and lung malt lymphoma
cured lymphoma not the autoimmune disease. Ann Rheum Dis
2001; 60: 174–176.
Marmont AM. Coincidental autoimmune diseases in patients
transplanted for conventional indications. Best Pract Res Clin
Haematol 2004; 17: 223–232.
Griffith LM, Pavletic SZ, Tyndall A, Bredeson CN, Bowen JD,
Childs RW et al. Feasability of allogeneic hematopoietic stem
cell transplantation for autoimmune disease: position statement from a National Institute of Allergy and Infectious
Diseases and National Cancer-Institute-sponsored International Workshop, Bethesda, MD, March 12 and 13, 2005. Biol
Blood Marrow Transplant 2005; 11: 862–870.
Sykes M, Nikolic B. Treatment of severe autoimmune disease
by stem-cell transplantation. Nature 2005; 435: 620–627.
Appelbaum FR. Hematopoietic stem cell transplantation as
immunotherapy. Nat Med 2001; 41: 385–389.
Slavin S, Nagler A, Varadi G, Or R. Graft vs autoimmunity
following allogeneic nonmyeloablative blood stem cell transplantation in a patient with chronic myelogenous leukemia and
severe systemic psoriasis and psoriatic polyarthritis. Exp
Hematol 2000; 28: 853–857.
Marmont AM. Is there any evidence of a graft-versusautoimmunity effect in allogeneic transplantation? Blood
Marrow Transplant Rev 2004; 4: 11.
Hinterberger W, Hinterberger-Fischer M, Marmont AM.
Clinically demonstrable anti-autoimmunity mediated by allogeneic immune cells favorably affects outcome after stem cell
transplantation in human autoimmune diseases. Bone Marrow
Transplant 2002; 30: 753–759.
Oyama Y, Papadopoulos EB, Mirando M, Traynor AE, Burt
RK. Allogeneic stem cell transplantation for Evans syndrome.
Bone Marrow Transplant 2001; 28: 903–905.
Li H, Kaufman CL, Boggs SS, Johnson PC, Patrene KD,
Ildstad ST. Mixed allogeneic chimerism induced by a sublethal
approach prevents autoimmune diabetes and reverses insulitis
in nonobese diabetic (NOD) mice. J Immunol 1996; 156:
380–387.
Seung E, Iwakoshi N, Woda BA, Markees TG, Mordes JP,
Rossini AA et al. Allogeneic hematopoietic chimerism in
mice treated with sublethal myeloablation and anti-CD154
antibody: absence of graft-versus-host disease, induction of
skin allograft tolerance, and prevention of recurrent auto-
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
immunity in islet-allografted NOD/Lt mice. Blood 2000; 95:
2175–2182.
Good RA, Wang BY, El-Badri NS, Steele A, Verjee T. Mixed
bone marrow or mixed stem cell transplantation for prevention
or treatment of lupus-like disease in mice. Exp Nephrol 2002;
10: 408–420.
Chakrabarti S, Handa SK, Byron RJ, Griffiths MJ, Milligan
DW. Will mixed chimerism cure autoimmune diseases after a
nonmyeloablative stem cell transplant? Transplantation 2001;
72: 340–342.
Burt RK, Oyama Y, Verda L, Onigley K, Brush M, Yaung K
et al. Induction of remission of severe and refractory
rheumatoid arthritis by allogeneic mixed chimerism. Arthritis
Rheum 2004; 50: 2466–2470.
Kojima R, Kami M, Kim S-W, Murashige N, Kishi Y, Hori A
et al. Induction of graft-versus-autoimmune (GVA) disease
effect against refractory psoriasis by complete donor-type
chimerism, and graft-versus-host disease after allogeneic
hematopoietic system cell transplantation. Bone Marrow
Transplant 2003; 32: 439–442.
Seifert B, Passweg JK, Heim D, Rovo A, Meyer-Monard S,
Bushner S et al. Complete remission of alopecia universalis
after allogeneic hematopoietic stem cell transplantation.
Transplantation 2005; 105: 426–427.
Marmont AM, Gualandi F, van Lint MT, Bacigalupo A.
Refractory Evans syndrome treated with allogeneic SCT
followed by DLI. Demonstration of a graft-versus autoimmunity effect. Bone Marrow Transplant 2003; 31: 399–401.
Musso M, Porretto F, Crescimanno A, Polizzi V, Scalone R.
Donor lymphocyte infusions for refractory pure red cell
aplasia relapsing after both autologous and allogeneic peripheral stem cell transplantation. Bone Marrow Transplant 2004;
33: 769–771.
Hayden PJ, Crampe M, Lawler M, Conneally E, McCann SR,
Vandenberghe E et al. Use of DLI to achieve complete donor
chimerism in a patient receiving systemic immunosuppression
for refractory AIHA post-NST. Bone Marrow Transplant 2005;
36: 735–736.
McKendry RJ, Huebsch L, Leclair B. Progression of
rheumatoid arthritis following bone marrow transplantation.
A case report with a 13-year follow-up. Arthritis Rheum 1996;
39: 1246–1253.
Tapprich C, Feuk R, Schneider P, Bernhardt A, Haas R,
Kobbe G. Early recurrence of rheumatoid arthritis after
nonmyeloablative allogeneic blood stem cell transplantation
in a patient with multiple myeloma. Bone Marrow Transplant
2003; 32: 629–631.
El-Badri NS, Maheshwari A, Sanberg PR. Mesenchymal stem
cells in autoimmune disease. Stem Cell Dev 2004; 13: 463–472.
Marmont AM, Gualandi F, van Lint MT, Bacigalupo A.
Refractory Evans syndrome treated with allogeneic SCT
followed by DLI. Demonstration of a graft-versus-autoimmunity effect. Bone Marrow Transplant 2003; 31: 399–402.
Marmont AM, Gualandi F, Occhini D, Morandi F, Ferretti E,
Pezzolo A et al. Catastrophic relapse of Evans syndrome five
years after allogeneic BMT notwithstanding full donor
chimerism Terminal haemolytic-uremic syndrome. Autoimmunity (in press).
Hoyer BE, Manz RA, Radbruch A, Hiepe F. Long-lived
plasma cells and their contribution to autoimmunity. Ann NY
Acad Sci 2005; 1050: 124–133.
Manz RA, Hanser AE, Hiepe F, Radbruch A. Maintenance of
serum antibody levels. Annu Rev Immunol 2005; 23: 367–386.
Hoyer B, Moser K, Hanser AE, Peddinghaus A, Voigt C, Eilat
D et al. Short-lived plasma blasts and long-lived plasma cells
contribute to chronic humoral autoimmunity in NZB/W mice.
J Exp Med 2004; 199: 1577–1584.
Bone Marrow Transplantation
Editorial
4
55 Marmont AM. Historical perspective and rationale of HSCT
for autoimmune diseases. In: Burt RK, Marmont AM (eds).
Stem Cell Therapy for Autoimmune Diseases. Landes
Bioscience: Georgetown, USA, 2004.
56 O’Connor BP, Raman VS, Erickson LD, Cook WJ, Weaver
LK, Ahonen C et al. BCMA is essential for the survival of
Bone Marrow Transplantation
long-lived bone marrow plasma cells. J Exp Med 2004; 199:
91–98.
57 Shapiro-Shelef M, Lin K-I, Savitsky D, Liao J,
Calame K. Blimp-1 is required for maintenance of longlived plasma cells in the bone marrow. J Exp Med 2005; 202:
1471–1476.